1
|
Rama E, Mohapatra SR, Sugimura Y, Suzuki T, Siebert S, Barmin R, Hermann J, Baier J, Rix A, Lemainque T, Koletnik S, Elshafei AS, Pallares RM, Dadfar SM, Tolba RH, Schulz V, Jankowski J, Apel C, Akhyari P, Jockenhoevel S, Kiessling F. In vitro and in vivo evaluation of biohybrid tissue-engineered vascular grafts with transformative 1H/ 19F MRI traceable scaffolds. Biomaterials 2024; 311:122669. [PMID: 38906013 DOI: 10.1016/j.biomaterials.2024.122669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 06/23/2024]
Abstract
Biohybrid tissue-engineered vascular grafts (TEVGs) promise long-term durability due to their ability to adapt to hosts' needs. However, the latter calls for sensitive non-invasive imaging approaches to longitudinally monitor their functionality, integrity, and positioning. Here, we present an imaging approach comprising the labeling of non-degradable and degradable TEVGs' components for their in vitro and in vivo monitoring by hybrid 1H/19F MRI. TEVGs (inner diameter 1.5 mm) consisted of biodegradable poly(lactic-co-glycolic acid) (PLGA) fibers passively incorporating superparamagnetic iron oxide nanoparticles (SPIONs), non-degradable polyvinylidene fluoride scaffolds labeled with highly fluorinated thermoplastic polyurethane (19F-TPU) fibers, a smooth muscle cells containing fibrin blend, and endothelial cells. 1H/19F MRI of TEVGs in bioreactors, and after subcutaneous and infrarenal implantation in rats, revealed that PLGA degradation could be faithfully monitored by the decreasing SPIONs signal. The 19F signal of 19F-TPU remained constant over weeks. PLGA degradation was compensated by cells' collagen and α-smooth-muscle-actin deposition. Interestingly, only TEVGs implanted on the abdominal aorta contained elastin. XTT and histology proved that our imaging markers did not influence extracellular matrix deposition and host immune reaction. This concept of non-invasive longitudinal assessment of cardiovascular implants using 1H/19F MRI might be applicable to various biohybrid tissue-engineered implants, facilitating their clinical translation.
Collapse
Affiliation(s)
- Elena Rama
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Saurav Ranjan Mohapatra
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Yukiharu Sugimura
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Tomoyuki Suzuki
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Siebert
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Roman Barmin
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Juliane Hermann
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Jasmin Baier
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Anne Rix
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Teresa Lemainque
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany; Department of Diagnostic and Interventional Radiology, Medical Faculty, RWTH Aachen University, 52074 Aachen, Germany
| | - Susanne Koletnik
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Asmaa Said Elshafei
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Roger Molto Pallares
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Seyed Mohammadali Dadfar
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany; Ardena Oss, 5349 AB Oss, the Netherlands
| | - René H Tolba
- Institute for Laboratory Animal Science and Experimental Surgery, Medical Faculty, RWTH Aachen International University, Aachen, Germany
| | - Volkmar Schulz
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), University of Maastricht, the Netherlands
| | - Christian Apel
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Payam Akhyari
- Department of Cardiac Surgery, Medical Faculty and RWTH University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles, AME-Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, Faculty of Medicine, RWTH Aachen University, Forckenbeckstraße 55, 52074 Aachen, Germany.
| |
Collapse
|
2
|
Chen Q, Wang C, Wang H, Xiao J, Zhou Y, Gu S, Xu W, Yang H. Strengthened Decellularized Porcine Valves via Polyvinyl Alcohol as a Template Improving Processability. Polymers (Basel) 2023; 16:16. [PMID: 38201681 PMCID: PMC10780456 DOI: 10.3390/polym16010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 01/12/2024] Open
Abstract
The heart valve is crucial for the human body, which directly affects the efficiency of blood transport and the normal functioning of all organs. Generally, decellularization is one method of tissue-engineered heart valve (TEHV), which can deteriorate the mechanical properties and eliminate allograft immunogenicity. In this study, removable polyvinyl alcohol (PVA) is used to encapsulate decellularized porcine heart valves (DHVs) as a dynamic template to improve the processability of DHVs, such as suturing. Mechanical tests show that the strength and elastic modulus of DHVs treated with different concentrations of PVA significantly improve. Without the PVA layer, the valve would shift during suture puncture and not achieve the desired suture result. The in vitro results indicate that decellularized valves treated with PVA can sustain the adhesion and growth of human umbilical vein endothelial cells (HUVECs). All results above show that the DHVs treated with water-soluble PVA have good mechanical properties and cytocompatibility to ensure post-treatment. On this basis, the improved processability of DHV treated with PVA enables a new paradigm for the manufacturing of scaffolds, making it easy to apply.
Collapse
Affiliation(s)
- Qingqing Chen
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (Q.C.); (C.W.); (Y.Z.); (S.G.)
| | - Chaorong Wang
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (Q.C.); (C.W.); (Y.Z.); (S.G.)
| | - Han Wang
- Key Laboratory of Green Processing and Functional New Textile Materials of Ministry of Education, Wuhan Textile University, Wuhan 430200, China (J.X.); (W.X.)
- Institute for Frontier Materials, Deakin University, Geelong, VIC 3216, Australia
| | - Jinfeng Xiao
- Key Laboratory of Green Processing and Functional New Textile Materials of Ministry of Education, Wuhan Textile University, Wuhan 430200, China (J.X.); (W.X.)
| | - Yingshan Zhou
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (Q.C.); (C.W.); (Y.Z.); (S.G.)
| | - Shaojin Gu
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (Q.C.); (C.W.); (Y.Z.); (S.G.)
| | - Weilin Xu
- Key Laboratory of Green Processing and Functional New Textile Materials of Ministry of Education, Wuhan Textile University, Wuhan 430200, China (J.X.); (W.X.)
| | - Hongjun Yang
- College of Materials Science and Engineering, Wuhan Textile University, Wuhan 430200, China; (Q.C.); (C.W.); (Y.Z.); (S.G.)
- Key Laboratory of Green Processing and Functional New Textile Materials of Ministry of Education, Wuhan Textile University, Wuhan 430200, China (J.X.); (W.X.)
| |
Collapse
|
3
|
Li MX, Wei QQ, Mo HL, Ren Y, Zhang W, Lu HJ, Joung YK. Challenges and advances in materials and fabrication technologies of small-diameter vascular grafts. Biomater Res 2023; 27:58. [PMID: 37291675 PMCID: PMC10251629 DOI: 10.1186/s40824-023-00399-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/21/2023] [Indexed: 06/10/2023] Open
Abstract
The arterial occlusive disease is one of the leading causes of cardiovascular diseases, often requiring revascularization. Lack of suitable small-diameter vascular grafts (SDVGs), infection, thrombosis, and intimal hyperplasia associated with synthetic vascular grafts lead to a low success rate of SDVGs (< 6 mm) transplantation in the clinical treatment of cardiovascular diseases. The development of fabrication technology along with vascular tissue engineering and regenerative medicine technology allows biological tissue-engineered vascular grafts to become living grafts, which can integrate, remodel, and repair the host vessels as well as respond to the surrounding mechanical and biochemical stimuli. Hence, they potentially alleviate the shortage of existing vascular grafts. This paper evaluates the current advanced fabrication technologies for SDVGs, including electrospinning, molding, 3D printing, decellularization, and so on. Various characteristics of synthetic polymers and surface modification methods are also introduced. In addition, it also provides interdisciplinary insights into the future of small-diameter prostheses and discusses vital factors and perspectives for developing such prostheses in clinical applications. We propose that the performance of SDVGs can be improved by integrating various technologies in the near future.
Collapse
Affiliation(s)
- Mei-Xian Li
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Qian-Qi Wei
- Department of Infectious Diseases, General Hospital of Tibet Military Command, Xizang, China
| | - Hui-Lin Mo
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
| | - Yu Ren
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China
- School of Textile and Clothing, Nantong University, Nantong, 226019, China
| | - Wei Zhang
- National and Local Joint Engineering Research Center of Technical Fiber Composites for Safety and Protection, Nantong University, Nantong, 226019, China.
- School of Textile and Clothing, Nantong University, Nantong, 226019, China.
| | - Huan-Jun Lu
- Institute of Special Environmental Medicine, Nantong University, Nantong, 226019, China.
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.
- Division of Bio-Medical Science and Technology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
4
|
Xu W, Yao M, He M, Chen S, Lu Q. Precise Preparation of a Multilayer Tubular Cell Sheet with Well-Aligned Cells in Different Layers to Simulate Native Arteries. ACS APPLIED MATERIALS & INTERFACES 2023; 15:19966-19975. [PMID: 37043742 DOI: 10.1021/acsami.3c00471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Compared with artificial vascular grafts, bottom-up tubular cell sheets (TCSs) without scaffolds have shown promise for patients with cardiovascular disease. However, TCS therapy also faces the challenges of lengthy maturation time, elaborate operation, and weak mechanical strength. In this work, a structured small-diameter vascular graft (SDVG), consisting of three layers of TCSs, with different cell types and arrangements, was fabricated using layer-by-layer assembly of naturally formed TCSs and further cell culture. To this end, a surface-patterned collagen-coated cylindrical substrate was designed for the efficient harvesting of naturally formed and well-aligned TCSs. The patterned collagen (type I) layer facilitated the adhesion and orientation of cells, and a continuous tubular cell monolayer was naturally formed after approximately 4 days in cell culture. Biocompatible near-infrared (NIR) light was used to trigger the photothermal phase transition of the collagen coated on the cylindrical substrate to dissociate the collagen layer. As a result, an intact TCS could be harvested within a few minutes. These naturally formed and well-aligned TCSs exhibited outstanding free-standing performance without rugosity, facilitating their operability and practical application. A ring tensile test showed that orientation was critical for improving the mechanical properties of TCSs. The layer-by-layer assembly of SDVGs not only is easy to manipulate and has a short preparation time but also overcomes the bottleneck of forming a hierarchically structured vascular graft. This approach shows promise for repairing damaged blood vessels.
Collapse
Affiliation(s)
- Wei Xu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, the State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240 China
| | - Mengting Yao
- School of Chemical Science and Engineering, Tongji University, Shanghai 200092 China
| | - Meng He
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, the State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240 China
| | - Shuangshuang Chen
- Institute of Translational Medicine, Shanghai University, Shanghai 200444 China
| | - Qinghua Lu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, the State Key Laboratory of Metal Matrix Composites, Shanghai Jiao Tong University, Shanghai 200240 China
| |
Collapse
|
5
|
Breuer T, Jimenez M, Humphrey JD, Shinoka T, Breuer CK. Tissue Engineering of Vascular Grafts: A Case Report From Bench to Bedside and Back. Arterioscler Thromb Vasc Biol 2023; 43:399-409. [PMID: 36633008 PMCID: PMC9974789 DOI: 10.1161/atvbaha.122.318236] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/29/2022] [Indexed: 01/13/2023]
Abstract
For over 25 years, our group has used regenerative medicine strategies to develop improved biomaterials for use in congenital heart surgery. Among other applications, we developed a tissue-engineered vascular graft (TEVG) by seeding tubular biodegradable polymeric scaffolds with autologous bone marrow-derived mononuclear cells. Results of our first-in-human study demonstrated feasibility as the TEVG transformed into a living vascular graft having an ability to grow, making it the first engineered graft with growth potential. Yet, outcomes of this first Food and Drug Administration-approved clinical trial evaluating safety revealed a prohibitively high incidence of early TEVG stenosis, preventing the widespread use of this promising technology. Mechanistic studies in mouse models provided important insight into the development of stenosis and enabled advanced computational models. Computational simulations suggested both a novel inflammation-driven, mechano-mediated process of in vivo TEVG development and an unexpected natural history, including spontaneous reversal of the stenosis. Based on these in vivo and in silico discoveries, we have been able to rationally design strategies for inhibiting TEVG stenosis that have been validated in preclinical large animal studies and translated to the clinic via a new Food and Drug Administration-approved clinical trial. This progress would not have been possible without the multidisciplinary approach, ranging from small to large animal models and computational simulations. This same process is expected to lead to further advances in scaffold design, and thus next generation TEVGs.
Collapse
Affiliation(s)
- Thomas Breuer
- Nationwide Children's Hospital, Columbus, OH (T.B., M.J., T.S., C.K.B.)
| | - Michael Jimenez
- Nationwide Children's Hospital, Columbus, OH (T.B., M.J., T.S., C.K.B.)
| | - Jay D Humphrey
- Yale University, School of Engineering and Applied Science, New Haven, CT (J.D.H.)
| | - Toshiharu Shinoka
- Nationwide Children's Hospital, Columbus, OH (T.B., M.J., T.S., C.K.B.)
| | | |
Collapse
|
6
|
Mirhaidari GJ, Barker JC, Breuer CK, Reinhardt JW. Implanted Tissue-Engineered Vascular Graft Cell Isolation with Single-Cell RNA Sequencing Analysis. Tissue Eng Part C Methods 2023; 29:72-84. [PMID: 36719780 PMCID: PMC9968626 DOI: 10.1089/ten.tec.2022.0189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 01/17/2023] [Indexed: 02/01/2023] Open
Abstract
The advent of single-cell RNA sequencing (scRNA-Seq) has brought with it the ability to gain greater insights into the cellular composition of tissues and heterogeneity in gene expression within specific cell types. For tissue-engineered blood vessels, this is particularly impactful to better understand how neotissue forms and remodels into tissue resembling a native vessel. A notable challenge, however, is the ability to separate cells from synthetic biomaterials to generate high-quality single-cell suspensions to interrogate the cellular composition of our tissue-engineered vascular grafts (TEVGs) during active remodeling in situ. We present here a simple, commercially available approach to separate cells within our TEVG from the residual scaffold for downstream use in a scRNA-Seq workflow. Utilizing this method, we identified the cell populations comprising explanted TEVGs and compared these with results from immunohistochemical analysis. The process began with explanted TEVGs undergoing traditional mechanical and enzymatic dissociation to separate cells from scaffold and extracellular matrix proteins. Magnetically labeled antibodies targeting murine origin cells were incubated with enzymatic digests of TEVGs containing cells and scaffold debris in suspension allowing for separation by utilizing a magnetic separator column. Single-cell suspensions were processed through 10 × Genomics and data were analyzed utilizing R to generate cell clusters. Expression data provided new insights into a diverse composition of phenotypically unique subclusters within the fibroblast, macrophage, smooth muscle cell, and endothelial cell populations contributing to the early neotissue remodeling stages of TEVGs. These populations were correlated qualitatively and quantitatively with immunohistochemistry highlighting for the first time the potential of scRNA-Seq to provide exquisite detail into the host cellular response to an implanted TEVG. These results additionally demonstrate magnetic cell isolation is an effective method for generating high-quality cell suspensions for scRNA-Seq. While this method was utilized for our group's TEVGs, it has broader applications to other implantable materials that use biodegradable synthetic materials as part of scaffold composition. Impact statement Single-cell RNA sequencing is an evolving technology with the ability to provide detailed information on the cellular composition of remodeling biomaterials in vivo. This present work details an effective approach for separating nondegraded biomaterials from cells for downstream RNA-sequencing analysis. We applied this method to implanted tissue-engineered vascular grafts and for the first time describe the cellular composition of the remodeling graft at a single-cell gene expression level. While this method was effective in our scaffold, it has broad applicability to other implanted biomaterials that necessitate separation of cell from residual scaffold materials for single-cell RNA sequencing.
Collapse
Affiliation(s)
- Gabriel J.M. Mirhaidari
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jenny C. Barker
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
- Department of Plastic and Reconstructive Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Christopher K. Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - James W. Reinhardt
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
7
|
Wang X, Chan V, Corridon PR. Acellular Tissue-Engineered Vascular Grafts from Polymers: Methods, Achievements, Characterization, and Challenges. Polymers (Basel) 2022; 14:4825. [PMID: 36432950 PMCID: PMC9695055 DOI: 10.3390/polym14224825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 11/11/2022] Open
Abstract
Extensive and permanent damage to the vasculature leading to different pathogenesis calls for developing innovative therapeutics, including drugs, medical devices, and cell therapies. Innovative strategies to engineer bioartificial/biomimetic vessels have been extensively exploited as an effective replacement for vessels that have seriously malfunctioned. However, further studies in polymer chemistry, additive manufacturing, and rapid prototyping are required to generate highly engineered vascular segments that can be effectively integrated into the existing vasculature of patients. One recently developed approach involves designing and fabricating acellular vessel equivalents from novel polymeric materials. This review aims to assess the design criteria, engineering factors, and innovative approaches for the fabrication and characterization of biomimetic macro- and micro-scale vessels. At the same time, the engineering correlation between the physical properties of the polymer and biological functionalities of multiscale acellular vascular segments are thoroughly elucidated. Moreover, several emerging characterization techniques for probing the mechanical properties of tissue-engineered vascular grafts are revealed. Finally, significant challenges to the clinical transformation of the highly promising engineered vessels derived from polymers are identified, and unique perspectives on future research directions are presented.
Collapse
Affiliation(s)
- Xinyu Wang
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Vincent Chan
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| | - Peter R. Corridon
- Department of Biomedical Engineering and Healthcare Engineering Innovation Center, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Department of Immunology and Physiology, College of Medicine and Health Sciences, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi P.O. Box 127788, United Arab Emirates
| |
Collapse
|
8
|
Wolf KJ, Weiss JD, Uzel SGM, Skylar-Scott MA, Lewis JA. Biomanufacturing human tissues via organ building blocks. Cell Stem Cell 2022; 29:667-677. [PMID: 35523137 PMCID: PMC9617289 DOI: 10.1016/j.stem.2022.04.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The construction of human organs on demand remains a tantalizing vision to solve the organ donor shortage. Yet, engineering tissues that recapitulate the cellular and architectural complexity of native organs is a grand challenge. The use of organ building blocks (OBBs) composed of multicellular spheroids, organoids, and assembloids offers an important pathway for creating organ-specific tissues with the desired cellular-to-tissue-level organization. Here, we review the differentiation, maturation, and 3D assembly of OBBs into functional human tissues and, ultimately, organs for therapeutic repair and replacement. We also highlight future challenges and areas of opportunity for this nascent field.
Collapse
Affiliation(s)
- Kayla J Wolf
- Wyss Institute for Biologically Inspired Engineering & John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, MA 02138, USA
| | - Jonathan D Weiss
- Department of Bioengineering, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA
| | - Sebastien G M Uzel
- Wyss Institute for Biologically Inspired Engineering & John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, MA 02138, USA
| | - Mark A Skylar-Scott
- Department of Bioengineering, Stanford University, 240 Pasteur Drive, Stanford, CA 94304, USA; BASE Initiative, Betty Irene Moore Children's Heart Center, Lucile Packard Children's Hospital, Stanford University School of Medicine, Stanford, CA 94304, USA.
| | - Jennifer A Lewis
- Wyss Institute for Biologically Inspired Engineering & John A. Paulson School of Engineering and Applied Sciences, Harvard University, 29 Oxford Street, Cambridge, MA 02138, USA.
| |
Collapse
|
9
|
Blum KM, Mirhaidari G, Breuer CK. Tissue engineering: Relevance to neonatal congenital heart disease. Semin Fetal Neonatal Med 2022; 27:101225. [PMID: 33674254 PMCID: PMC8390581 DOI: 10.1016/j.siny.2021.101225] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Congenital heart disease (CHD) represents a large clinical burden, representing the most common cause of birth defect-related death in the newborn. The mainstay of treatment for CHD remains palliative surgery using prosthetic vascular grafts and valves. These devices have limited effectiveness in pediatric patients due to thrombosis, infection, limited endothelialization, and a lack of growth potential. Tissue engineering has shown promise in providing new solutions for pediatric CHD patients through the development of tissue engineered vascular grafts, heart patches, and heart valves. In this review, we examine the current surgical treatments for congenital heart disease and the research being conducted to create tissue engineered products for these patients. While much research remains to be done before tissue engineering becomes a mainstay of clinical treatment for CHD patients, developments have been progressing rapidly towards translation of tissue engineering devices to the clinic.
Collapse
Affiliation(s)
- Kevin M Blum
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus, OH, USA; Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA.
| | - Gabriel Mirhaidari
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus OH, USA,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus OH, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, The Abigail Wexner Research Institute, Nationwide Childrens Hospital, Columbus, OH, USA.
| |
Collapse
|
10
|
Lorentz KL, Gupta P, Shehabeldin MS, Cunnane EM, Ramaswamy AK, Verdelis K, DiLeo MV, Little SR, Weinbaum JS, Sfeir CS, Mandal BB, Vorp DA. CCL2 loaded microparticles promote acute patency in silk-based vascular grafts implanted in rat aortae. Acta Biomater 2021; 135:126-138. [PMID: 34496284 PMCID: PMC8595801 DOI: 10.1016/j.actbio.2021.08.049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 08/04/2021] [Accepted: 08/27/2021] [Indexed: 01/22/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide, often associated with coronary artery occlusion. A common intervention for arterial blockage utilizes a vascular graft to bypass the diseased artery and restore downstream blood flow; however, current clinical options exhibit high long-term failure rates. Our goal was to develop an off-the-shelf tissue-engineered vascular graft capable of delivering a biological payload based on the monocyte recruitment factor C-C motif chemokine ligand 2 (CCL2) to induce remodeling. Bi-layered silk scaffolds consisting of an inner porous and outer electrospun layer were fabricated using a custom blend of Antherea Assama and Bombyx Mori silk (lyogel). Lyogel silk scaffolds alone (LG), and lyogel silk scaffolds containing microparticles (LGMP) were tested. The microparticles (MPs) were loaded with either CCL2 (LGMP+) or water (LGMP-). Scaffolds were implanted as abdominal aortic interposition grafts in Lewis rats for 1 and 8 weeks. 1-week implants exhibited patency rates of 50% (7/14), 100% (10/10), and 100% (5/5) in the LGMP-, LGMP+, and LG groups, respectively. The significantly higher patency rate for the LGMP+ group compared to the LGMP- group (p = 0.0188) suggests that CCL2 can prevent acute occlusion. Immunostaining of the explants revealed a significantly higher density of macrophages (CD68+ cells) within the outer vs. inner layer of LGMP- and LGMP+ constructs but not in LG constructs. After 8 weeks, there were no significant differences in patency rates between groups. All patent scaffolds at 8 weeks showed signs of remodeling; however, stenosis was observed within the majority of explants. This study demonstrated the successful fabrication of a custom blended silk scaffold functionalized with cell-mimicking microparticles to facilitate controlled delivery of a biological payload improving their in vivo performance. STATEMENT OF SIGNIFICANCE: This study outlines the development of a custom blended silk-based tissue-engineered vascular graft (TEVG) for use in arterial bypass or replacement surgery. A custom mixture of silk was formulated to improve biocompatibility and cellular binding to the tubular scaffold. Many current approaches to TEVGs include cells that encourage graft cellularization and remodeling; however, our technology incorporates a microparticle based delivery platform capable of delivering bioactive molecules that can mimic the function of seeded cells. In this study, we load the TEVGs with microparticles containing a monocyte attractant and demonstrate improved performance in terms of unobstructed blood flow versus blank microparticles. The acellular nature of this technology potentially reduces risk, increases reproducibility, and results in a more cost-effective graft when compared to cell-based options.
Collapse
Affiliation(s)
- Katherine L Lorentz
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Prerak Gupta
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India
| | - Mostafa S Shehabeldin
- Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA; Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, United States; Department of Periodontics and Preventive Dentistry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Eoghan M Cunnane
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; Tissue Engineering Research Group, Dept. of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Aneesh K Ramaswamy
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States
| | - Konstantinos Verdelis
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, United States
| | - Morgan V DiLeo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Steven R Little
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justin S Weinbaum
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Pathology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Charles S Sfeir
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Oral and Craniofacial Sciences, University of Pittsburgh, Pittsburgh, PA; Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, United States; Department of Periodontics and Preventive Dentistry, University of Pittsburgh, Pittsburgh, PA, United States
| | - Biman B Mandal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, India; Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, India; School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, India.
| | - David A Vorp
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States; Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, PA, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, United States; Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA, United States; Center for Vascular Remodeling and Regeneration, University of Pittsburgh, Pittsburgh, PA, United States; The Clinical & Translational Sciences Institute, University of Pittsburgh, Pittsburgh, PA, United States; Department of Mechanical Engineering and Materials Science, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
11
|
Schwarz EL, Kelly JM, Blum KM, Hor KN, Yates AR, Zbinden JC, Verma A, Lindsey SE, Ramachandra AB, Szafron JM, Humphrey JD, Shin'oka T, Marsden AL, Breuer CK. Hemodynamic performance of tissue-engineered vascular grafts in Fontan patients. NPJ Regen Med 2021; 6:38. [PMID: 34294733 PMCID: PMC8298568 DOI: 10.1038/s41536-021-00148-w] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 06/11/2021] [Indexed: 02/06/2023] Open
Abstract
In the field of congenital heart surgery, tissue-engineered vascular grafts (TEVGs) are a promising alternative to traditionally used synthetic grafts. Our group has pioneered the use of TEVGs as a conduit between the inferior vena cava and the pulmonary arteries in the Fontan operation. The natural history of graft remodeling and its effect on hemodynamic performance has not been well characterized. In this study, we provide a detailed analysis of the first U.S. clinical trial evaluating TEVGs in the treatment of congenital heart disease. We show two distinct phases of graft remodeling: an early phase distinguished by rapid changes in graft geometry and a second phase of sustained growth and decreased graft stiffness. Using clinically informed and patient-specific computational fluid dynamics (CFD) simulations, we demonstrate how changes to TEVG geometry, thickness, and stiffness affect patient hemodynamics. We show that metrics of patient hemodynamics remain within normal ranges despite clinically observed levels of graft narrowing. These insights strengthen the continued clinical evaluation of this technology while supporting recent indications that reversible graft narrowing can be well tolerated, thus suggesting caution before intervening clinically.
Collapse
Affiliation(s)
- Erica L Schwarz
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
| | - John M Kelly
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kevin M Blum
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Kan N Hor
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Andrew R Yates
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Jacob C Zbinden
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Biomedical Engineering, The Ohio State University, Columbus, OH, USA
| | - Aekaansh Verma
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Stephanie E Lindsey
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | | | - Jason M Szafron
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Toshiharu Shin'oka
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
- Department of Cardiothoracic Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Alison L Marsden
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
- Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
- Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
12
|
Biomimetic tubular scaffold with heparin conjugation for rapid degradation in in situ regeneration of a small diameter neoartery. Biomaterials 2021; 274:120874. [PMID: 34051629 DOI: 10.1016/j.biomaterials.2021.120874] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 01/22/2023]
Abstract
To address the clinical need for readily available small diameter vascular grafts, biomimetic tubular scaffolds were developed for rapid in situ blood vessel regeneration. The tubular scaffolds were designed to have an inner layer that is porous, interconnected, and with a nanofibrous architecture, which provided an excellent microenvironment for host cell invasion and proliferation. Through the synthesis of poly(spirolactic-co-lactic acid) (PSLA), a highly functional polymer with a norbornene substituting a methyl group in poly(l-lactic acid) (PLLA), we were able to covalently attach biomolecules onto the polymer backbone via thiol-ene click chemistry to impart desirable functionalities to the tubular scaffolds. Specifically, heparin was conjugated on the scaffolds in order to prevent thrombosis when implanted in situ. By controlling the amount of covalently attached heparin we were able to modulate the physical properties of the tubular scaffold, resulting in tunable wettability and degradation rate while retaining the porous and nanofibrous morphology. The scaffolds were successfully tested as rat abdominal aortic replacements. Patency and viability were confirmed through dynamic ultrasound and histological analysis of the regenerated tissue. The harvested tissue showed excellent vascular cellular infiltration, proliferation, and migration with laminar cellular arrangement. Furthermore, we achieved both complete reendothelialization of the vessel lumen and native-like media extracellular matrix. No signs of aneurysm or hyperplasia were observed after 3 months of vessel replacement. Taken together, we have developed an effective vascular graft able to generate small diameter blood vessels that can function in a rat model.
Collapse
|
13
|
Liu X, Yan J, Liu J, Wang Y, Yin J, Fu J. Fabrication of a dual-layer cell-laden tubular scaffold for nerve regeneration and bile duct reconstruction. Biofabrication 2021; 13. [PMID: 33873178 DOI: 10.1088/1758-5090/abf995] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/19/2021] [Indexed: 01/29/2023]
Abstract
Tubular scaffolds serve as a controllable extracellular environment to guide the repair and regeneration of tissues. But it is still a challenge to achieve both excellent mechanical properties and cell compatibility of artificial scaffolds for long-term structural and biological stability. In this study, a four-step solution casting method was developed to fabricate dual-layer cell-laden tubular scaffolds for nerve and bile duct regeneration. The dual-layer tubular scaffold consisted of a bone marrow mesenchymal stem cells (BMSCs)-laden hydrogel inner layer and an outer layer of gelatin methacrylate (GelMA)/polyethylene glycol diacrylate. While the inner layer had a good biocompatibility, the outer layer had desired mechanical properties. The interfacial toughness, Young's modulus, maximum tensile strain, and compressive modulus of dual-layer tubular scaffolds were 65 J m-2, 122.37 ± 23.21 kPa, 100.87 ± 40.10%, and 39.14 ± 18.56 N m-1, respectively. More importantly, the fabrication procedure was very cell-friendly, since the BMSC viability encapsulated in the inner layer of 10% (w/v) GelMA reached 94.68 ± 0.43% after 5 d of culture. Then, a preliminary evaluation of the potential application of dual-layer tubular scaffolds as nerve conduits and biliary scaffolds was performed, and demonstrated that the cell-laden dual-layer tubular scaffolds proposed in this work are expected to extend the application of tubular scaffolds in tissue engineering.
Collapse
Affiliation(s)
- Xixia Liu
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China.,School of Mechanical Engineering, Guizhou University, Guiyang 550025, People's Republic of China
| | - Jianing Yan
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou 310016, People's Republic of China.,Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, People's Republic of China.,Zhejiang University Innovation Center of Minimally Invasive Technology and Medical Equipment, Hangzhou 310016, People's Republic of China
| | - Jingyi Liu
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China
| | - Yifan Wang
- Department of General Surgery, Sir Run Run Shaw Hospital Affiliated to School of Medicine, Zhejiang University, Hangzhou 310016, People's Republic of China.,Zhejiang Provincial Key Laboratory of Laparoscopic Technology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, People's Republic of China.,Zhejiang Province Medical Research Center of Minimally Invasive Diagnosis and Treatment of Abdominal Diseases, Hangzhou 310016, People's Republic of China
| | - Jun Yin
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China
| | - Jianzhong Fu
- The State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China.,Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou 310028, People's Republic of China
| |
Collapse
|
14
|
Khosravi R, Ramachandra AB, Szafron JM, Schiavazzi DE, Breuer CK, Humphrey JD. A computational bio-chemo-mechanical model of in vivo tissue-engineered vascular graft development. Integr Biol (Camb) 2021; 12:47-63. [PMID: 32222759 DOI: 10.1093/intbio/zyaa004] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 01/26/2020] [Accepted: 02/04/2020] [Indexed: 12/15/2022]
Abstract
Stenosis is the primary complication of current tissue-engineered vascular grafts used in pediatric congenital cardiac surgery. Murine models provide considerable insight into the possible mechanisms underlying this situation, but they are not efficient for identifying optimal changes in scaffold design or therapeutic strategies to prevent narrowing. In contrast, computational modeling promises to enable time- and cost-efficient examinations of factors leading to narrowing. Whereas past models have been limited by their phenomenological basis, we present a new mechanistic model that integrates molecular- and cellular-driven immuno- and mechano-mediated contributions to in vivo neotissue development within implanted polymeric scaffolds. Model parameters are inferred directly from in vivo measurements for an inferior vena cava interposition graft model in the mouse that are augmented by data from the literature. By complementing Bayesian estimation with identifiability analysis and simplex optimization, we found optimal parameter values that match model outputs with experimental targets and quantify variability due to measurement uncertainty. Utility is illustrated by parametrically exploring possible graft narrowing as a function of scaffold pore size, macrophage activity, and the immunomodulatory cytokine transforming growth factor beta 1 (TGF-β1). The model captures salient temporal profiles of infiltrating immune and synthetic cells and associated secretion of cytokines, proteases, and matrix constituents throughout neovessel evolution, and parametric studies suggest that modulating scaffold immunogenicity with early immunomodulatory therapies may reduce graft narrowing without compromising compliance.
Collapse
Affiliation(s)
- Ramak Khosravi
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | | | - Jason M Szafron
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Daniele E Schiavazzi
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN, USA
| | - Christopher K Breuer
- Center for Regenerative Medicine, Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA.,Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
15
|
Zheng M, Guo J, Li Q, Yang J, Han Y, Yang H, Yu M, Zhong L, Lu D, Li L, Sun L. Syntheses and characterization of anti-thrombotic and anti-oxidative Gastrodin-modified polyurethane for vascular tissue engineering. Bioact Mater 2021; 6:404-419. [PMID: 32995669 PMCID: PMC7486448 DOI: 10.1016/j.bioactmat.2020.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
Vascular grafts must avoid negative inflammatory responses and thrombogenesis to prohibit fibrotic deposition immediately upon implantation and promote the regeneration of small diameter blood vessels (<6 mm inner diameter). Here, polyurethane (PU) elastomers incorporating anti-coagulative and anti-inflammatory Gastrodin were fabricated. The films had inter-connected pores with porosities equal to or greater than 86% and pore sizes ranging from 250 to 400 μm. Incorporation of Gastrodin into PU films resulted in desirable mechanical properties, hydrophilicity, swelling ratios and degradation rates without collapse. The released Gastrodin maintained bioactivity over 21 days as assessed by its anti-oxidative capability. The Gastrodin/PU had better anti-coagulation response (less observable BSA, fibrinogen and platelet adhesion/activation and suppressed clotting in whole blood). Red blood cell compatibility, measured by hemolysis, was greatly improved with 2Gastrodin/PU compared to other Gastrodin/PU groups. Notably, Gastrodin/PU upregulated anti-oxidant factors Nrf2 and HO-1 expression in H2O2 treated HUVECs, correlated with decreasing pro-inflammatory cytokines TNF-α and IL-1β in RAW 264.7 cells. Upon implantation in a subcutaneous pocket, PU was encapsulated by an obvious fibrous capsule, concurrent with a large amount of inflammatory cell infiltration, while Gastrodin/PU induced a thinner fibrous capsule, especially 2Gastrodin/PU. Further, enhanced adhesion and proliferation of HUVECs seeded onto films in vitro demonstrated that 2Gastrodin/PU could help cell recruitment, as evidenced by rapid host cell infiltration and substantial blood vessel formation in vivo. These results indicate that 2Gastrodin/PU has the potential to facilitate blood vessel regeneration, thus providing new insight into the development of clinically effective vascular grafts.
Collapse
Affiliation(s)
- Meng Zheng
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jiazhi Guo
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Qing Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Jian Yang
- Department of Biomedical Engineering, Materials Research Institute, The Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Yi Han
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Hongcai Yang
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Mali Yu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lianmei Zhong
- Department of Neurology, The First Affiliated Hospital, Kunming Medical University, Kunming, 650500, China
| | - Di Lu
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Limei Li
- Yunnan Key Laboratory of Stem Cell and Regenerative Medicine, Science and Technology Achievement Incubation Center, Kunming Medical University, Kunming, 650500, China
| | - Lin Sun
- Department of Cardiology, The Second Affiliated Hospital, Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
16
|
Mallis P, Kostakis A, Stavropoulos-Giokas C, Michalopoulos E. Future Perspectives in Small-Diameter Vascular Graft Engineering. Bioengineering (Basel) 2020; 7:160. [PMID: 33321830 PMCID: PMC7763104 DOI: 10.3390/bioengineering7040160] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 12/04/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
The increased demands of small-diameter vascular grafts (SDVGs) globally has forced the scientific society to explore alternative strategies utilizing the tissue engineering approaches. Cardiovascular disease (CVD) comprises one of the most lethal groups of non-communicable disorders worldwide. It has been estimated that in Europe, the healthcare cost for the administration of CVD is more than 169 billion €. Common manifestations involve the narrowing or occlusion of blood vessels. The replacement of damaged vessels with autologous grafts represents one of the applied therapeutic approaches in CVD. However, significant drawbacks are accompanying the above procedure; therefore, the exploration of alternative vessel sources must be performed. Engineered SDVGs can be produced through the utilization of non-degradable/degradable and naturally derived materials. Decellularized vessels represent also an alternative valuable source for the development of SDVGs. In this review, a great number of SDVG engineering approaches will be highlighted. Importantly, the state-of-the-art methodologies, which are currently employed, will be comprehensively presented. A discussion summarizing the key marks and the future perspectives of SDVG engineering will be included in this review. Taking into consideration the increased number of patients with CVD, SDVG engineering may assist significantly in cardiovascular reconstructive surgery and, therefore, the overall improvement of patients' life.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Alkiviadis Kostakis
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece;
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| |
Collapse
|
17
|
Mirhaidari GJ, Barker JC, Zbinden JC, Santantonio BM, Chang YC, Best CA, Reinhardt JW, Blum KM, Yi T, Breuer CK. Tissue Engineered Vascular Graft Recipient Interleukin 10 Status Is Critical for Preventing Thrombosis. Adv Healthc Mater 2020; 9:e2001094. [PMID: 33073543 PMCID: PMC7936649 DOI: 10.1002/adhm.202001094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 09/28/2020] [Indexed: 01/07/2023]
Abstract
Tissue engineered vascular grafts (TEVGs) are a promising technology, but are hindered by occlusion. Seeding with bone-marrow derived mononuclear cells (BM-MNCs) mitigates occlusion, yet the precise mechanism remains unclear. Seeded cells disappear quickly and potentially mediate an anti-inflammatory effect through paracrine signaling. Here, a series of reciprocal genetic TEVG implantations plus recombinant protein treatment is reported to investigate what role interleukin-10, an anti-inflammatory cytokine, plays from both host and seeded cells. TEVGs seeded with BM-MNCs from wild-type and IL-10 KO mice, plus unseeded grafts, are implanted into wild-type and IL-10 KO mice. Wild-type mice with unseeded grafts also receive recombinant IL-10. Serial ultrasound evaluates occlusion and TEVGs are harvested at 14 d for immunohistochemical analysis. TEVGs in IL-10 KO mice have significantly higher occlusion incidence compared to wild-type mice attributed to acute (<3 d) thrombosis. Cell seeding rescues TEVGs in IL-10 KO mice comparable to wild-type patency. IL-10 from the host and seeded cells do not significantly influence graft inflammation and macrophage phenotype, yet IL-10 treatment shows interesting biologic effects including decreasing cell proliferation and increasing M2 macrophage polarization. IL-10 from the host is critical for preventing TEVG thrombosis and seeded BM-MNCs exert a significant anti-thrombotic effect in IL-10 KO mice.
Collapse
Affiliation(s)
- Gabriel J.M. Mirhaidari
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Jenny C. Barker
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Jacob C. Zbinden
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Brevan M. Santantonio
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Yu-Chun Chang
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Cameron A. Best
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - James W. Reinhardt
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Kevin M. Blum
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Tai Yi
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| | - Christopher K. Breuer
- The Abigail Wexner Research Institute at Nationwide Children’s Hospital, 575 Children’s Crossroad, Research III, WB4160 A1, Columbus, OH, 43215, United States of America
| |
Collapse
|
18
|
Yang L, Li X, Wu Y, Du P, Sun L, Yu Z, Song S, Yin J, Ma X, Jing C, Zhao J, Chen H, Dong Y, Zhang Q, Zhao L. Preparation of PU/Fibrin Vascular Scaffold with Good Biomechanical Properties and Evaluation of Its Performance in vitro and in vivo. Int J Nanomedicine 2020; 15:8697-8715. [PMID: 33192062 PMCID: PMC7656973 DOI: 10.2147/ijn.s274459] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/25/2020] [Indexed: 01/22/2023] Open
Abstract
PURPOSE The development of tissue-engineered blood vessels provides a new source of donors for coronary artery bypass grafting and peripheral blood vessel transplantation. Fibrin fiber has good biocompatibility and is an ideal tissue engineering vascular scaffold, but its mechanical property needs improvement. METHODS We mixed polyurethane (PU) and fibrin to prepare the PU/fibrin vascular scaffolds by using electrospinning technology in order to enhance the mechanical properties of fibrin scaffold. We investigated the morphological, mechanical strength, hydrophilicity, degradation, blood and cell compatibility of PU/fibrin (0:100), PU/fibrin (5:95), PU/fibrin (15:85) and PU/fibrin (25:75) vascular scaffolds. Based on the results in vitro, PU/fibrin (15:85) was selected for transplantation in vivo to repair vascular defects, and the extracellular matrix formation, vascular remodeling, and immune response were evaluated. RESULTS The results indicated that the fiber diameter of the PU/fibrin (15:85) scaffold was about 712nm. With the increase of PU content, the mechanical strength of the composite scaffolds increased, however, the degradation rate decreased gradually. The PU/fibrin scaffold showed good hydrophilicity and hemocompatibility. PU/fibrin (15:85) vascular scaffold could promote the adhesion and proliferation of mesenchymal stromal cells (MSCs). Quantitative RT-PCR experimental results showed that the expression of collagen, survivin and vimentin genes in PU/fibrin (15:85) was higher than that in PU/fibrin (25:75). The results in vivo indicated the mechanical properties and compliance of PU/fibrin grafts could meet clinical requirements and the proportion of thrombosis or occlusion was significantly lower. The graft showed strong vasomotor response, and the smooth muscle cells, endothelial cells, and ECM deposition of the neoartery were comparable to that of native artery after 3 months. At 3 months, the amount of macrophages in PU/fibrin grafts was significantly lower, and the secretion of pro-inflammatory and anti-inflammatory cytokines decreased. CONCLUSION PU/fibrin (15:85) vascular scaffolds had great potential to be used as small-diameter tissue engineering blood vessels.
Collapse
Affiliation(s)
- Lei Yang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Xiafei Li
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yiting Wu
- Xiacun Community Health Service Center, Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, People’s Republic of China
| | - Pengchong Du
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Department of Cardio-Thoracic Surgery, Third Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Lulu Sun
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Zhenyang Yu
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Shuang Song
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Jianshen Yin
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Xianfen Ma
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Changqin Jing
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Junqiang Zhao
- College of Medical Engineering, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Hongli Chen
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Yuzhen Dong
- Department of Orthopedics, First Affiliated Hospital, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Qiqing Zhang
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
| | - Liang Zhao
- College of Life Science and Technology, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Key Laboratory of Cardiac Structure Research, Zhengzhou Seventh People’s Hospital, Zhengzhou, People’s Republic of China
- The Central Lab, The Third People’s Hospital of Datong, Datong, People’s Republic of China
| |
Collapse
|
19
|
Next-generation tissue-engineered heart valves with repair, remodelling and regeneration capacity. Nat Rev Cardiol 2020; 18:92-116. [PMID: 32908285 DOI: 10.1038/s41569-020-0422-8] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2020] [Indexed: 02/06/2023]
Abstract
Valvular heart disease is a major cause of morbidity and mortality worldwide. Surgical valve repair or replacement has been the standard of care for patients with valvular heart disease for many decades, but transcatheter heart valve therapy has revolutionized the field in the past 15 years. However, despite the tremendous technical evolution of transcatheter heart valves, to date, the clinically available heart valve prostheses for surgical and transcatheter replacement have considerable limitations. The design of next-generation tissue-engineered heart valves (TEHVs) with repair, remodelling and regenerative capacity can address these limitations, and TEHVs could become a promising therapeutic alternative for patients with valvular disease. In this Review, we present a comprehensive overview of current clinically adopted heart valve replacement options, with a focus on transcatheter prostheses. We discuss the various concepts of heart valve tissue engineering underlying the design of next-generation TEHVs, focusing on off-the-shelf technologies. We also summarize the latest preclinical and clinical evidence for the use of these TEHVs and describe the current scientific, regulatory and clinical challenges associated with the safe and broad clinical translation of this technology.
Collapse
|
20
|
Toong DWY, Toh HW, Ng JCK, Wong PEH, Leo HL, Venkatraman S, Tan LP, Ang HY, Huang Y. Bioresorbable Polymeric Scaffold in Cardiovascular Applications. Int J Mol Sci 2020; 21:E3444. [PMID: 32414114 PMCID: PMC7279389 DOI: 10.3390/ijms21103444] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/06/2020] [Accepted: 05/08/2020] [Indexed: 12/12/2022] Open
Abstract
Advances in material science and innovative medical technologies have allowed the development of less invasive interventional procedures for deploying implant devices, including scaffolds for cardiac tissue engineering. Biodegradable materials (e.g., resorbable polymers) are employed in devices that are only needed for a transient period. In the case of coronary stents, the device is only required for 6-8 months before positive remodelling takes place. Hence, biodegradable polymeric stents have been considered to promote this positive remodelling and eliminate the issue of permanent caging of the vessel. In tissue engineering, the role of the scaffold is to support favourable cell-scaffold interaction to stimulate formation of functional tissue. The ideal outcome is for the cells to produce their own extracellular matrix over time and eventually replace the implanted scaffold or tissue engineered construct. Synthetic biodegradable polymers are the favoured candidates as scaffolds, because their degradation rates can be manipulated over a broad time scale, and they may be functionalised easily. This review presents an overview of coronary heart disease, the limitations of current interventions and how biomaterials can be used to potentially circumvent these shortcomings in bioresorbable stents, vascular grafts and cardiac patches. The material specifications, type of polymers used, current progress and future challenges for each application will be discussed in this manuscript.
Collapse
Affiliation(s)
- Daniel Wee Yee Toong
- School of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore 639798, Singapore; (D.W.Y.T.); (L.P.T.)
| | - Han Wei Toh
- National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore; (H.W.T.); (J.C.K.N.); (P.E.H.W.)
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore;
| | - Jaryl Chen Koon Ng
- National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore; (H.W.T.); (J.C.K.N.); (P.E.H.W.)
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore;
| | - Philip En Hou Wong
- National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore; (H.W.T.); (J.C.K.N.); (P.E.H.W.)
- Duke-NUS Medical School, National University of Singapore, 8 College Road, Singapore 169857, Singapore
| | - Hwa Liang Leo
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore;
| | - Subramanian Venkatraman
- Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117575, Singapore;
| | - Lay Poh Tan
- School of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore 639798, Singapore; (D.W.Y.T.); (L.P.T.)
| | - Hui Ying Ang
- National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore; (H.W.T.); (J.C.K.N.); (P.E.H.W.)
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore 117583, Singapore;
| | - Yingying Huang
- School of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore 639798, Singapore; (D.W.Y.T.); (L.P.T.)
| |
Collapse
|
21
|
Lee W, Hong Y, Dai G. 3D bioprinting of vascular conduits for pediatric congenital heart repairs. Transl Res 2019; 211:35-45. [PMID: 31034816 PMCID: PMC6702035 DOI: 10.1016/j.trsl.2019.03.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 03/21/2019] [Accepted: 03/24/2019] [Indexed: 12/27/2022]
Abstract
In children with congenital heart defects, surgical correction often involves the use of valves, patches or vascular conduits to establish anatomic continuity. Due to the differences between the pediatric and adult populations, tissue reconstruction in pediatric patients requires a substantially different approach from those in adults. Cardiovascular anatomy of children with congenital heart defect vary, which requires tailored surgical operations for each patient. Since grafts used in these palliative surgeries are sensitive to the local hemodynamic environments, their geometries need to be precisely designed to ensure long-term performance. Tissue engineered vascular grafts (TEVGs) have made tremendous progress over the past decade, but it remains difficult to fabricate patient- and operation-specific vascular grafts. This review summarizes historical milestones of TEVG development for repairing pediatric congenital defects and current clinical outcomes. We also highlight ongoing works on 3D bioprinting of TEVGs with complex geometries and address the current limitations of each technique. Although 3D bioprinted vascular grafts with appropriate functions are yet to be developed, some of the current researches are promising to create better patient specific tissue engineered vascular grafts in the future.
Collapse
Affiliation(s)
- Wenhan Lee
- Department of Bioengineering, Northeastern University, Boston, Massachusetts
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Guohao Dai
- Department of Bioengineering, Northeastern University, Boston, Massachusetts.
| |
Collapse
|
22
|
Best CA, Szafron JM, Rocco KA, Zbinden J, Dean EW, Maxfield MW, Kurobe H, Tara S, Bagi PS, Udelsman BV, Khosravi R, Yi T, Shinoka T, Humphrey JD, Breuer CK. Differential outcomes of venous and arterial tissue engineered vascular grafts highlight the importance of coupling long-term implantation studies with computational modeling. Acta Biomater 2019; 94:183-194. [PMID: 31200116 PMCID: PMC6819998 DOI: 10.1016/j.actbio.2019.05.063] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 05/07/2019] [Accepted: 05/24/2019] [Indexed: 12/13/2022]
Abstract
Electrospinning is commonly used to generate polymeric scaffolds for tissue engineering. Using this approach, we developed a small-diameter tissue engineered vascular graft (TEVG) composed of poly-ε-caprolactone-co-l-lactic acid (PCLA) fibers and longitudinally assessed its performance within both the venous and arterial circulations of immunodeficient (SCID/bg) mice. Based on in vitro analysis demonstrating complete loss of graft strength by 12 weeks, we evaluated neovessel formation in vivo over 6-, 12- and 24-week periods. Mid-term observations indicated physiologic graft function, characterized by 100% patency and luminal matching with adjoining native vessel in both the venous and arterial circulations. An active and robust remodeling process was characterized by a confluent endothelial cell monolayer, macrophage infiltrate, and extracellular matrix deposition and remodeling. Long-term follow-up of venous TEVGs at 24 weeks revealed viable neovessel formation beyond graft degradation when implanted in this high flow, low-pressure environment. Arterial TEVGs experienced catastrophic graft failure due to aneurysmal dilatation and rupture after 14 weeks. Scaffold parameters such as porosity, fiber diameter, and degradation rate informed a previously described computational model of vascular growth and remodeling, and simulations predicted the gross differential performance of the venous and arterial TEVGs over the 24-week time course. Taken together, these results highlight the requirement for in vivo implantation studies to extend past the critical time period of polymer degradation, the importance of differential neotissue deposition relative to the mechanical (pressure) environment, and further support the utility of predictive modeling in the design, use, and evaluation of TEVGs in vivo. STATEMENT OF SIGNIFICANCE: Herein, we apply a biodegradable electrospun vascular graft to the arterial and venous circulations of the mouse and follow recipients beyond the point of polymer degradation. While venous implants formed viable neovessels, arterial grafts experienced catastrophic rupture due to aneurysmal dilation. We then inform a previously developed computational model of tissue engineered vascular graft growth and remodeling with parameters specific to the electrospun scaffolds utilized in this study. Remarkably, model simulations predict the differential performance of the venous and arterial constructs over 24 weeks. We conclude that computational simulations should inform the rational selection of scaffold parameters to fabricate tissue engineered vascular grafts that must be followed in vivo over time courses extending beyond polymer degradation.
Collapse
Affiliation(s)
- Cameron A Best
- Center for Regenerative Medicine, Tissue Engineering Program, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, United States.
| | - Jason M Szafron
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | | | - Jacob Zbinden
- Center for Regenerative Medicine, Tissue Engineering Program, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Biomedical Engineering Graduate Program, The Ohio State University College of Engineering, Columbus, OH, United States
| | - Ethan W Dean
- Department of Orthopaedic Surgery, University of Florida, Gainesville, FL, United States
| | - Mark W Maxfield
- Department of Thoracic Surgery, University of Massachusetts Memorial Medical Center, Worcester, MA, United States
| | - Hirotsugu Kurobe
- Department of Cardiovascular Surgery, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Shuhei Tara
- Department of Cardiovascular Medicine, Nippon Medical School, Tokyo, Japan
| | - Paul S Bagi
- Department of Orthopaedic Surgery, Yale-New Haven Hospital, New Haven, CT, United States
| | - Brooks V Udelsman
- Department of Surgery, Massachusetts General Hospital, Boston, MA, United States
| | - Ramak Khosravi
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| | - Tai Yi
- Center for Regenerative Medicine, Tissue Engineering Program, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, Tissue Engineering Program, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Department of Cardiac Surgery, Nationwide Children's Hospital, Columbus, OH, United States
| | - Jay D Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, United States
| | - Christopher K Breuer
- Center for Regenerative Medicine, Tissue Engineering Program, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Department of Surgery, Nationwide Children's Hospital, Columbus, OH, United States
| |
Collapse
|
23
|
Arora S, Yim EKF, Toh YC. Environmental Specification of Pluripotent Stem Cell Derived Endothelial Cells Toward Arterial and Venous Subtypes. Front Bioeng Biotechnol 2019; 7:143. [PMID: 31259171 PMCID: PMC6587665 DOI: 10.3389/fbioe.2019.00143] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/28/2019] [Indexed: 12/25/2022] Open
Abstract
Endothelial cells (ECs) are required for a multitude of cardiovascular clinical applications, such as revascularization of ischemic tissues or endothelialization of tissue engineered grafts. Patient derived primary ECs are limited in number, have donor variabilities and their in vitro phenotypes and functions can deteriorate over time. This necessitates the exploration of alternative EC sources. Although there has been a recent surge in the use of pluripotent stem cell derived endothelial cells (PSC-ECs) for various cardiovascular clinical applications, current differentiation protocols yield a heterogeneous EC population, where their specification into arterial or venous subtypes is undefined. Since arterial and venous ECs are phenotypically and functionally different, inappropriate matching of exogenous ECs to host sites can potentially affect clinical efficacy, as exemplified by venous graft mismatch when placed into an arterial environment. Therefore, there is a need to design and employ environmental cues that can effectively modulate PSC-ECs into a more homogeneous arterial or venous phenotype for better adaptation to the host environment, which will in turn contribute to better application efficacy. In this review, we will first give an overview of the developmental and functional differences between arterial and venous ECs. This provides the foundation for our subsequent discussion on the different bioengineering strategies that have been investigated to varying extent in providing biochemical and biophysical environmental cues to mature PSC-ECs into arterial or venous subtypes. The ability to efficiently leverage on a combination of biochemical and biophysical environmental cues to modulate intrinsic arterio-venous specification programs in ECs will greatly facilitate future translational applications of PSC-ECs. Since the development and maintenance of arterial and venous ECs in vivo occur in disparate physio-chemical microenvironments, it is conceivable that the application of these environmental factors in customized combinations or magnitudes can be used to selectively mature PSC-ECs into an arterial or venous subtype.
Collapse
Affiliation(s)
- Seep Arora
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore
| | - Evelyn K F Yim
- Department of Chemical Engineering, University of Waterloo, Waterloo, ON, Canada
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, Singapore, Singapore.,Singapore Institute for Neurotechnology (SINAPSE), National University of Singapore, Singapore, Singapore.,Biomedical Institute for Global Health Research and Technology (BIGHEART), National University of Singapore, Singapore, Singapore.,NUS Tissue Engineering Program, National University of Singapore, Singapore, Singapore
| |
Collapse
|
24
|
Regenerative and durable small-diameter graft as an arterial conduit. Proc Natl Acad Sci U S A 2019; 116:12710-12719. [PMID: 31182572 DOI: 10.1073/pnas.1905966116] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Despite significant research efforts, clinical practice for arterial bypass surgery has been stagnant, and engineered grafts continue to face postimplantation challenges. Here, we describe the development and application of a durable small-diameter vascular graft with tailored regenerative capacity. We fabricated small-diameter vascular grafts by electrospinning fibrin tubes and poly(ε-caprolactone) fibrous sheaths, which improved suture retention strength and enabled long-term survival. Using surface topography in a hollow fibrin microfiber tube, we enable immediate, controlled perfusion and formation of a confluent endothelium within 3-4 days in vitro with human endothelial colony-forming cells, but a stable endothelium is noticeable at 4 weeks in vivo. Implantation of acellular or endothelialized fibrin grafts with an external ultrathin poly(ε-caprolactone) sheath as an interposition graft in the abdominal aorta of a severe combined immunodeficient Beige mouse model supports normal blood flow and vessel patency for 24 weeks. Mechanical properties of the implanted grafts closely approximate the native abdominal aorta properties after just 1 week in vivo. Fibrin mediated cellular remodeling, stable tunica intima and media formation, and abundant matrix deposition with organized collagen layers and wavy elastin lamellae. Endothelialized grafts evidenced controlled healthy remodeling with delayed and reduced macrophage infiltration alongside neo vasa vasorum-like structure formation, reduced calcification, and accelerated tunica media formation. Our studies establish a small-diameter graft that is fabricated in less than 1 week, mediates neotissue formation and incorporation into the native tissue, and matches the native vessel size and mechanical properties, overcoming main challenges in arterial bypass surgery.
Collapse
|
25
|
Wu Y, Yu C, Xing M, Wang L, Guan G. Surface modification of polyvinyl alcohol (PVA)/polyacrylamide (PAAm) hydrogels with polydopamine and REDV for improved applicability. J Biomed Mater Res B Appl Biomater 2019; 108:117-127. [PMID: 30912304 DOI: 10.1002/jbm.b.34371] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 03/01/2019] [Accepted: 03/05/2019] [Indexed: 02/03/2023]
Abstract
Developing a small-diameter vascular graft with a satisfactory performance in terms of mechanical and biological properties remains a challenging issue because of comprehensive requirements from clinical applications. Polyvinyl alcohol (PVA)/polyacrylamide (PAAm) hydrogels exhibit many desirable characteristics for small-diameter vascular grafts because of their tunable mechanical properties, especially high compliance. However, poor cells adhesion hinders their application for endothelialization in situ. Therefore, in the present work, polydopamine (PDA) and tetrapeptide Arg-Glu-Asp-Val (REDV) were used to functionalize the hydrogels surface and improve cells adhesion. A series of characterizations were systematically conducted to examine the applicability of coated hydrogels to small-diameter vascular grafts. Results showed that bare and coated hydrogels have appropriate structural stability, and no significant differences in tensile properties could be found after being coated with PDA or PDA-REDV. The hydrophilicity of the hydrogels decreased with the coatings of PDA and especially PDA-REDV to improve protein adsorption, porcine iliac artery endothelial cells (PIECs) adhesion, viability, proliferation, and spreading on the hydrogels. Lower hemolysis percentages and higher blood clotting index values were attained for the hydrogels, suggesting their satisfactory hemocompatibility. Overall, the present work provided insights into the development of a novel hydrogel-based small-diameter vascular graft. © 2019 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 108B:117-127, 2020.
Collapse
Affiliation(s)
- Yufen Wu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Chenglong Yu
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Meiyi Xing
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Lu Wang
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| | - Guoping Guan
- Key Laboratory of Textile Science & Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai, 201620, China
| |
Collapse
|
26
|
Deryabin P, Griukova A, Shatrova A, Petukhov A, Nikolsky N, Borodkina A. Optimization of lentiviral transduction parameters and its application for CRISPR-based secretome modification of human endometrial mesenchymal stem cells. Cell Cycle 2019; 18:742-758. [PMID: 30880567 PMCID: PMC6464586 DOI: 10.1080/15384101.2019.1593650] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 02/08/2019] [Accepted: 02/19/2019] [Indexed: 12/31/2022] Open
Abstract
Mesenchymal stem cells (MSCs) hold a great promise for successful development of regenerative medicine. Among the plenty of uncovered MSCs sources, desquamated endometrium collected from the menstrual blood probably remains the most accessible. Though numerous studies have been published on human endometrium-derived mesenchymal stem cells (hMESCs) properties in the past years, there are only a few data regarding their genetic modulation. Moreover, there is a lack of information about the fate of the transduced hMESCs. The present study aimed to optimize hMESCs transduction parameters and apply Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 technology for genome and secretome modification. The fate of hMESCs transduced either in presence of polybrene (Pb) or protamine sulfate (Ps) was assessed by alterations in CD expression profile, growth rate, cell size, migration capability, osteogenic, adipogenic, and decidual differential potentials. Here, we postulated that the use of Ps for hMESCs genetic manipulations is preferable, as it has no impact on the stem-cell properties, whereas Pb application is undesirable, as it induces cellular senescence. Plasminogen activator inhibitor-1 was selected for further targeted hMESCs genome and secretome modification using CRISPR/Cas9 systems. The obtained data provide optimized transduction scheme for hMESCs and verification of its effectiveness by successful hMESCs genome editing via CRISPR/Cas9 technology.
Collapse
Affiliation(s)
- Pavel Deryabin
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Anastasiia Griukova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Alla Shatrova
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | - Alexey Petukhov
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Nikolay Nikolsky
- Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg, Russia
| | | |
Collapse
|
27
|
Birru B, Mekala NK, Parcha SR. Mechanistic role of perfusion culture on bone regeneration. J Biosci 2019; 44:23. [PMID: 30837374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Bone tissue engineering (BTE) aims to develop engineered bone tissue to substitute conventional bone grafts. To achieve this, culturing the cells on the biocompatible three-dimensional (3D) scaffold is one alternative approach. The new functional bone tissue regeneration could be feasible by the synergetic combinations of cells, biomaterials and bioreactors. Although the field of biomaterial design/development for BTE applications attained reasonable success, development of suitable bioreactor remains still a major challenge. Tissue engineering bioreactors provide the microenvironment required for neo-tissue regeneration, and also can be used to study the physio-chemical cues effect on cell proliferation and differentiation in order to produce functional tissue. In this direction, various bioreactors have been developed and evaluated for the successful development of engineered bone tissue. Continues assessment of tissue development and limitations of the bioreactors lead to the progression of perfusion flow bioreactor system. Improvements in perfusion reactor system were able to yield multiple tissue engineered constructs with uniform cell distribution, easy to operate protocols and also effectively handled for the functional tissue development to meet the adequate supply of engineered graft for clinical application.
Collapse
Affiliation(s)
- Bhaskar Birru
- Department of Biotechnology, National Institute of Technology, Warangal 506 004, TS, India
| | | | | |
Collapse
|
28
|
|
29
|
Shafiq M, Wang L, Zhi D, Zhang Q, Wang K, Wang L, Kim DH, Kong D, Kim SH. In situ blood vessel regeneration using neuropeptide substance P-conjugated small-diameter vascular grafts. J Biomed Mater Res B Appl Biomater 2018; 107:1669-1683. [PMID: 30315717 DOI: 10.1002/jbm.b.34260] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 09/18/2018] [Accepted: 09/23/2018] [Indexed: 12/16/2022]
Abstract
In situ blood vessel regeneration through host stem/progenitor cell mobilization may hold great promise for vascular reconstruction. Neuropeptide substance P (SP) has been shown to accelerate tissue repair by endogenous cell mobilization and recruitment. This study was aimed to evaluate the vascular regeneration potential of SP and heparin co-tethered vascular grafts. Polycaprolactone (PCL), PCL/SP-conjugated poly(L-lactide-co-ε-caprolactone) (PLCL-SP) (SP), and PCL/PLCL-SP/heparin-conjugated PLCL (Hep/SP) vascular grafts were implanted as rat abdominal aorta substitutes for up to 2 weeks and 4 weeks. Ex vivo results delineate that heparin can improve the hemocompatibility and SP can recruit mesenchymal stem cells. Histological and immunohistochemical staining reveal higher cellular infiltration and homogeneous cell distribution in SP and Hep/SP grafts than that of the control grafts. At 4 weeks, SP and Hep/SP grafts show the presence of cobblestone-like cells on the luminal side, whereas the surface of PCL grafts remains bare. Immunoflourescence staining using von Willibrand factor (vWF) antibody shows improved endothelialization in SP and Hep/SP grafts compared with the PCL grafts. SP and Hep/SP grafts also exhibit more numbers of α-smooth muscle actin-positive cells and laminin+ blood vessels than that of the control group. Evaluation of inflammatory response reveals that three groups did not differ in terms of the numbers of CD68+ macrophages, whereas SP and Hep/SP grafts show higher numbers of CD206+ macrophages. These results indicate that SP can induce endogenous tissue regeneration in cell-free grafts, which may be of great interest for regenerative medicine and tissue engineering applications. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1669-1683, 2019.
Collapse
Affiliation(s)
- Muhammad Shafiq
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.,State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China.,Center for Tissue Engineering & Regenerative Medicine, Department of Chemistry, Pakistan Institute of Engineering & Applied Sciences (PIEAS), Nilore, Islamabad, 45650, Pakistan
| | - Lina Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Dengke Zhi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Qiuying Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Lianyong Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Dong-Hwee Kim
- Department of Nano-Bio-Information Technology (NBIT), KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 136-701, Republic of Korea
| | - Deling Kong
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of Ministry of Education, College of Life Science, Nankai University, Tianjin, 300071, China
| | - Soo Hyun Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea.,Department of Nano-Bio-Information Technology (NBIT), KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 136-701, Republic of Korea
| |
Collapse
|
30
|
Human Pluripotent Stem Cells to Engineer Blood Vessels. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2018; 163:147-168. [PMID: 29090328 DOI: 10.1007/10_2017_28] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Development of pluripotent stem cells (PSCs) is a remarkable scientific advancement that allows scientists to harness the power of regenerative medicine for potential treatment of disease using unaffected cells. PSCs provide a unique opportunity to study and combat cardiovascular diseases, which continue to claim the lives of thousands each day. Here, we discuss the differentiation of PSCs into vascular cells, investigation of the functional capabilities of the derived cells, and their utilization to engineer microvascular beds or vascular grafts for clinical application. Graphical Abstract Human iPSCs generated from patients are differentiated toward ECs and perivascular cells for use in disease modeling, microvascular bed development, or vascular graft fabrication.
Collapse
|
31
|
Mechanical and structural analysis of the pulmonary valve in congenital heart defects: A presentation of two case studies. J Mech Behav Biomed Mater 2018; 89:9-12. [PMID: 30236979 DOI: 10.1016/j.jmbbm.2018.08.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/31/2018] [Accepted: 08/31/2018] [Indexed: 01/09/2023]
Abstract
OBJECTIVE Congenital Heart Disease (CHD) is the leading cause of pediatric mortality, with many cases affecting the right ventricular outflow tract (RVOT) or pulmonary valve (PV). Understanding the mechanics of the disease condition can provide insight into development of durable repair techniques and bioengineered replacement devices. This work presents a mechanical and structural analysis of the pulmonary valve of two pediatric cases. METHODS Two PV tissues were excised as part of the operative procedure. One PV was obtained from a 9-month-old with Noonan syndrome (Patient 1) and the other from a 6-month-old with tricuspid atresia (Patient 2). The leaflets were subjected to planar biaxial tensile testing and second harmonic generation (SHG) imaging for mechanical and structural evaluation. RESULTS AND DISCUSSION Patient 1 exhibited a more anisotropic mechanical response than Patient 2, with sample stiffness on par with that of adult PV tissue. Additionally, both samples showed radial and circumferential alignment of collagen fibers on the ventricularis and fibrosa sides of the leaflets, respectively. Collagen fibers on the fibrosa side were also more crimped than on the ventricularis side.
Collapse
|
32
|
Stowell CET, Wang Y. Quickening: Translational design of resorbable synthetic vascular grafts. Biomaterials 2018; 173:71-86. [PMID: 29772461 PMCID: PMC6492619 DOI: 10.1016/j.biomaterials.2018.05.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/19/2018] [Accepted: 05/03/2018] [Indexed: 12/17/2022]
Abstract
Traditional tissue-engineered vascular grafts have yet to gain wide clinical use. The difficulty of scaling production of these cell- or biologic-based products has hindered commercialization. In situ tissue engineering bypasses such logistical challenges by using acellular resorbable scaffolds. Upon implant, the scaffolds become remodeled by host cells. This review describes the scientific and translational advantages of acellular, synthetic vascular grafts. It surveys in vivo results obtained with acellular synthetics over their fifty years of technological development. Finally, it discusses emerging principles, highlights strategic considerations for designers, and identifies questions needing additional research.
Collapse
Affiliation(s)
| | - Yadong Wang
- Meinig School of Biomedical Engineering, Cornell University, USA.
| |
Collapse
|
33
|
Dzobo K, Thomford NE, Senthebane DA, Shipanga H, Rowe A, Dandara C, Pillay M, Motaung KSCM. Advances in Regenerative Medicine and Tissue Engineering: Innovation and Transformation of Medicine. Stem Cells Int 2018; 2018:2495848. [PMID: 30154861 PMCID: PMC6091336 DOI: 10.1155/2018/2495848] [Citation(s) in RCA: 219] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/22/2018] [Accepted: 07/08/2018] [Indexed: 02/08/2023] Open
Abstract
Humans and animals lose tissues and organs due to congenital defects, trauma, and diseases. The human body has a low regenerative potential as opposed to the urodele amphibians commonly referred to as salamanders. Globally, millions of people would benefit immensely if tissues and organs can be replaced on demand. Traditionally, transplantation of intact tissues and organs has been the bedrock to replace damaged and diseased parts of the body. The sole reliance on transplantation has created a waiting list of people requiring donated tissues and organs, and generally, supply cannot meet the demand. The total cost to society in terms of caring for patients with failing organs and debilitating diseases is enormous. Scientists and clinicians, motivated by the need to develop safe and reliable sources of tissues and organs, have been improving therapies and technologies that can regenerate tissues and in some cases create new tissues altogether. Tissue engineering and/or regenerative medicine are fields of life science employing both engineering and biological principles to create new tissues and organs and to promote the regeneration of damaged or diseased tissues and organs. Major advances and innovations are being made in the fields of tissue engineering and regenerative medicine and have a huge impact on three-dimensional bioprinting (3D bioprinting) of tissues and organs. 3D bioprinting holds great promise for artificial tissue and organ bioprinting, thereby revolutionizing the field of regenerative medicine. This review discusses how recent advances in the field of regenerative medicine and tissue engineering can improve 3D bioprinting and vice versa. Several challenges must be overcome in the application of 3D bioprinting before this disruptive technology is widely used to create organotypic constructs for regenerative medicine.
Collapse
Affiliation(s)
- Kevin Dzobo
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Dimakatso Alice Senthebane
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Hendrina Shipanga
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Arielle Rowe
- Cape Town Component, International Centre for Genetic Engineering and Biotechnology (ICGEB) and UCT Medical Campus, Wernher and Beit Building (South), Anzio Road, Observatory 7925, Cape Town, South Africa
| | - Collet Dandara
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular medicine, Faculty of Health Sciences, University of Cape Town, Observatory 7925, Cape Town, South Africa
| | - Michael Pillay
- Department of Biotechnology, Faculty of Applied and Computer Sciences, Vaal University of Technology, Vanderbijlpark 1900, South Africa
| | | |
Collapse
|
34
|
Optimization of oxygen transport within a tissue engineered vascular graft model using embedded micro-channels inspired by vasa vasorum. Chem Eng Sci 2018. [DOI: 10.1016/j.ces.2018.02.044] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
35
|
Abstract
The therapeutic replacement of diseased tubular tissue is hindered by the availability and suitability of current donor, autologous and synthetically derived protheses. Artificially created, tissue engineered, constructs have the potential to alleviate these concerns with reduced autoimmune response, high anatomical accuracy, long-term patency and growth potential. The advent of 3D bioprinting technology has further supplemented the technological toolbox, opening up new biofabrication research opportunities and expanding the therapeutic potential of the field. In this review, we highlight the challenges facing those seeking to create artificial tubular tissue with its associated complex macro- and microscopic architecture. Current biofabrication approaches, including 3D printing techniques, are reviewed and future directions suggested.
Collapse
|
36
|
Best C, Strouse R, Hor K, Pepper V, Tipton A, Kelly J, Shinoka T, Breuer C. Toward a patient-specific tissue engineered vascular graft. J Tissue Eng 2018; 9:2041731418764709. [PMID: 29568478 PMCID: PMC5858675 DOI: 10.1177/2041731418764709] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Accepted: 02/20/2018] [Indexed: 12/15/2022] Open
Abstract
Integrating three-dimensional printing with the creation of tissue-engineered vascular grafts could provide a readily available, patient-specific, autologous tissue source that could significantly improve outcomes in newborns with congenital heart disease. Here, we present the recent case of a candidate for our tissue-engineered vascular graft clinical trial deemed ineligible due to complex anatomical requirements and consider the application of three-dimensional printing technologies for a patient-specific graft. We 3D-printed a closed-disposable seeding device and validated that it performed equivalently to the traditional open seeding technique using ovine bone marrow–derived mononuclear cells. Next, our candidate’s preoperative imaging was reviewed to propose a patient-specific graft. A seeding apparatus was then designed to accommodate the custom graft and 3D-printed on a commodity fused deposition modeler. This exploratory feasibility study represents an important proof of concept advancing progress toward a rationally designed patient-specific tissue-engineered vascular graft for clinical application.
Collapse
Affiliation(s)
- Cameron Best
- Center for Regenerative Medicine, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.,Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Robert Strouse
- Research Innovation and Solutions, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA
| | - Kan Hor
- Department of Cardiology, The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Victoria Pepper
- Center for Regenerative Medicine, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| | - Amy Tipton
- Advanced Cardiac Imaging Laboratory, The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - John Kelly
- Center for Regenerative Medicine, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Cardiology, The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Toshiharu Shinoka
- Center for Regenerative Medicine, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Cardiothoracic Surgery, The Heart Center, Nationwide Children's Hospital, Columbus, OH, USA
| | - Christopher Breuer
- Center for Regenerative Medicine, The Research Institute, Nationwide Children's Hospital, Columbus, OH, USA.,Department of Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
37
|
Onwuka E, King N, Heuer E, Breuer C. The Heart and Great Vessels. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a031922. [PMID: 28289246 DOI: 10.1101/cshperspect.a031922] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardiovascular disease is the leading cause of mortality worldwide. We have made large strides over the past few decades in management, but definitive therapeutic options to address this health-care burden are still limited. Given the ever-increasing need, much effort has been spent creating engineered tissue to replaced diseased tissue. This article gives a general overview of this work as it pertains to the development of great vessels, myocardium, and heart valves. In each area, we focus on currently studied methods, limitations, and areas for future study.
Collapse
Affiliation(s)
- Ekene Onwuka
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Nakesha King
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Eric Heuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205
| | - Christopher Breuer
- Tissue Engineering and Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205.,College of Medicine, The Ohio State University, Columbus, Ohio 43210.,Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, Ohio 43205
| |
Collapse
|
38
|
Le V, Lee J, Chaterji S, Spencer A, Liu YL, Kim P, Yeh HC, Kim DH, Baker AB. Syndecan-1 in mechanosensing of nanotopological cues in engineered materials. Biomaterials 2018; 155:13-24. [PMID: 29156422 PMCID: PMC5738284 DOI: 10.1016/j.biomaterials.2017.11.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2017] [Accepted: 11/07/2017] [Indexed: 12/25/2022]
Abstract
The cells of the vascular system are highly sensitive to biophysical cues from their local cellular microenvironment. To engineer improved materials for vascular devices and delivery of cell therapies, a key challenge is to understand the mechanisms that cells use to sense biophysical cues from their environment. Syndecans are heparan sulfate proteoglycans (HSPGs) that consist of a protein core modified with heparan sulfate glycosaminoglycan chains. Due to their presence on the cell surface and their interaction with cytoskeletal and focal adhesion associated molecules, cell surface proteoglycans are well poised to serve as mechanosensors of the cellular microenvironment. Nanotopological cues have become recognized as major regulators of cell growth, migration and phenotype. We hypothesized that syndecan-1 could serve as a mechanosensor for nanotopological cues and can mediate the responsiveness of vascular smooth muscle cells to nanoengineered materials. We created engineered substrates made of polyurethane acrylate with nanogrooves using ultraviolet-assisted capillary force lithography. We cultured vascular smooth muscle cells with knockout of syndecan-1 on engineered substrates with varying compliance and nanotopology. We found that knockout of syndecan-1 reduced alignment of vascular smooth muscle cells to the nanogrooves under inflammatory treatments. In addition, we found that loss of syndecan-1 increased nuclear localization of Yap/Taz and phospho-Smad2/3 in response to nanogrooves. Syndecan-1 knockout vascular smooth muscle cells also had elevated levels of Rho-associated protein kinase-1 (Rock1), leading to increased cell stiffness and an enhanced contractile state in the cells. Together, our findings support that syndecan-1 knockout leads to alterations in mechanosensing of nanotopographical cues through alterations of in rho-associated signaling pathways, cell mechanics and mediators of the Hippo and TGF-β signaling pathways.
Collapse
Affiliation(s)
- Victoria Le
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Jason Lee
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Somali Chaterji
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Adrianne Spencer
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Yen-Liang Liu
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Peter Kim
- University of Washington, Department of Bioengineering, Seattle, WA, USA
| | - Hsin-Chih Yeh
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA
| | - Deok-Ho Kim
- University of Washington, Department of Bioengineering, Seattle, WA, USA
| | - Aaron B Baker
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, USA; Institute for Biomaterials, Drug Delivery and Regenerative Medicine, University of Texas at Austin, Austin, TX, USA; Institute for Computational Engineering and Sciences, University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
39
|
Recent Progress in Vascular Tissue-Engineered Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1064:123-144. [PMID: 30471030 DOI: 10.1007/978-981-13-0445-3_8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cardiovascular disease is the number one cause of death in the U.S and results in the loss of approximately one million lives and more than 400 billion U.S. dollars for treatments every year. Recently, tissue engineered blood vessels have been studied and developed as promising replacements for treatment with autologous veins. Here, we summarize the cell sources and methods to make tissue-engineered blood vessels (TEBVs), the recent progress in TEBV related research, and also the recent progress in TEBV related clinical studies.
Collapse
|
40
|
Jaganathan SK, Mani MP, Ayyar M, Krishnasamy NP, Nageswaran G. Blood compatibility and physicochemical assessment of novel nanocomposite comprising polyurethane and dietary carotino oil for cardiac tissue engineering applications. J Appl Polym Sci 2017. [DOI: 10.1002/app.45691] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Affiliation(s)
- Saravana Kumar Jaganathan
- Department for Management of Science and Technology Development; Ton Duc Thang University; Ho Chi Minh City Vietnam
- Faculty of Applied Sciences; Ton Duc Thang University; Ho Chi Minh City Vietnam
- IJNUTM Cardiovascular Engineering Centre, Department of Clinical Sciences, Faculty of Biosciences and Medical Engineering; Universiti Teknologi Malaysia; Skudai 81300 Johor Malaysia
| | - Mohan Prasath Mani
- IJNUTM Cardiovascular Engineering Centre, Department of Clinical Sciences, Faculty of Biosciences and Medical Engineering; Universiti Teknologi Malaysia; Skudai 81300 Johor Malaysia
| | - Manikandan Ayyar
- Department of Chemistry; Bharath Institute of Higher Education and Research; Bharath University Chennai 600073 Tamil Nadu India
| | | | - Gomathi Nageswaran
- Department of Chemistry; Indian Institute of Space Science and Technology; Trivandrum 695547 Kerala India
| |
Collapse
|
41
|
Jaganathan SK, M MP, Fauzi Ismail A, A M, N G. Production and hemocompatibility assessment of novel electrospun polyurethane nanofibers loaded with dietary virgin coconut oil for vascular graft applications. J BIOACT COMPAT POL 2017. [DOI: 10.1177/0883911517720815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
To develop biodegradable polymer scaffolds suitable for vascular tissue engineering applications, the bioengineering community has invested an extensive effort. The most common cause for the failure of vascular graft scaffolds is thrombosis. In this work, the scaffold based on polyurethane and virgin coconut oil was produced by electrospinning process for vascular tissue engineering applications with improved antithrombogenicity. The diameter of this electrospun polyurethane/virgin coconut oil composite was found to be reduced in the range of 886 ± 207 nm compared to pristine polyurethane which was in the range of 969 ± 217 nm. The Fourier transform infrared spectroscopy analysis revealed the interaction between polyurethane and virgin coconut oil as indicated by phase shifting of CH bond along with the formation of hydrogen bond. The contact angle measurement of fabricated composites was found to be increased owing to hydrophobic nature and also exhibited enhanced thermal stability as noted in thermogravimetric analysis. The atomic force microscopy analysis insinuated the increased surface roughness of the composite in comparison with the pure polyurethane. Developed scaffold resulted in delayed blood clotting as revealed by activated partial thromboplastin time and partial thromboplastin time assay. The hemolytic index of fabricated composites was found to be low indicating the enhanced safety of red blood cells. Hence, the newly developed nanofibrous composite scaffold could open the door for a suitable alternative for vascular graft applications.
Collapse
Affiliation(s)
- Saravana Kumar Jaganathan
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
- IJN-UTM Cardiovascular Engineering Center, Department of Clinical Sciences, Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai 81300, Johor, Malaysia
| | - Mohan Prasath M
- Faculty of Biosciences and Medical Engineering, Universiti Teknologi Malaysia, Skudai 81300, Johor, Malaysia
| | - Ahmad Fauzi Ismail
- Advanced Membrane Technology Research Centre (AMTEC), Universiti Teknologi Malaysia, Skudai 81310, Johor, Malaysia
| | - Manikandan A
- Department of Chemistry, Bharath Institute of Higher Education and Research, Bharath University Chennai 600073, Tamil Nadu, India
| | - Gomathi N
- Department of Chemistry, Indian Institute of Space Science and Technology, Thiruvananthapuram 695547, India
| |
Collapse
|
42
|
Wang Z, Wu Y, Wang J, Zhang C, Yan H, Zhu M, Wang K, Li C, Xu Q, Kong D. Effect of Resveratrol on Modulation of Endothelial Cells and Macrophages for Rapid Vascular Regeneration from Electrospun Poly(ε-caprolactone) Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2017; 9:19541-19551. [PMID: 28539044 DOI: 10.1021/acsami.6b16573] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Rapid endothelialization is a key factor that determines the success of small-diameter vascular grafts as an artery substitute in the treatment of cardiovascular diseases. Aimed to facilitate vascular regeneration, we developed a vascular scaffold loaded with resveratrol, which is a natural compound extracted from plants and showed multifaceted effects in cardiovascular protection. The tubular poly(ε-caprolactone) (PCL) scaffold was prepared by electrospinning with resveratrol in the PCL solution. In vitro assay demonstrated that resveratrol could be released from the scaffolds in a sustained and controlled manner. Cell culture results indicated that the migration of endothelial cells (ECs), nitric oxide production, and the ability of tube formation increased in the resveratrol-containing PCL scaffold groups compared with the PCL control. Meanwhile, the level of tumor necrosis factor (TNF)-α, the main proinflammatory factor secreted from macrophages, was reduced, and the messenger RNA expressions of the M2 macrophage-related genes were increased in the resveratrol-containing group. Further, in vivo implantation was performed by replacing rat abdominal aorta. We observed fast endothelialization and enhanced vascular regeneration in rats with resveratrol-containing scaffolds. The presence of resveratrol also induced a large number of M2 macrophages to infiltrate into the graft wall. Taken together, the incorporation of resveratrol into the PCL grafts enhanced the vascular regeneration by modulation of ECs and macrophages.
Collapse
Affiliation(s)
- Zhihong Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192, China
| | - Yifan Wu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Jianing Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Chuangnian Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192, China
| | - Hongyu Yan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Meifeng Zhu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Kai Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| | - Chen Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192, China
| | - Qingbo Xu
- Cardiovascular Division, King's College London BHF Centre , London SE5 9NU, U.K
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College , Tianjin 300192, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Science, Nankai University , Tianjin 300071, China
| |
Collapse
|
43
|
Shojaee M, Bashur CA. Compositions Including Synthetic and Natural Blends for Integration and Structural Integrity: Engineered for Different Vascular Graft Applications. Adv Healthc Mater 2017; 6. [PMID: 28371505 DOI: 10.1002/adhm.201700001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 02/13/2017] [Indexed: 11/07/2022]
Abstract
Tissue engineering approaches for small-diameter arteries require a scaffold that simultaneously maintains patency by preventing thrombosis and intimal hyperplasia, maintains its structural integrity after grafting, and allows integration. While synthetic and extracellular matrix-derived materials can provide some of these properties individually, developing a scaffold that provides the balanced properties needed for vascular graft survival in the clinic has been particularly challenging. After 30 years of research, there are now several scaffolds currently in clinical trials. However, these products are either being investigated for large-diameter applications or they require pre-seeding of endothelial cells. This progress report identifies important challenges unique to engineering vascular grafts for high pressure arteries less than 4 mm in diameter (e.g., coronary artery), and discusses limitations with the current usage of the term "small-diameter." Next, the composition and processing techniques used for generating tissue engineered vascular grafts (TEVGs) are discussed, with a focus on the benefits of blended materials. Other scaffolds for non-tissue engineering approaches and stents are also briefly mentioned for comparison. Overall, this progress report discusses the importance of defining the most critical challenges for small diameter TEVGs, developing new scaffolds to provide these properties, and determining acceptable benchmarks for scaffold responses in the body.
Collapse
|
44
|
Kajbafzadeh AM, Khorramirouz R, Kameli SM, Hashemi J, Bagheri A. Decellularization of Human Internal Mammary Artery: Biomechanical Properties and Histopathological Evaluation. Biores Open Access 2017; 6:74-84. [PMID: 28736690 PMCID: PMC5515095 DOI: 10.1089/biores.2016.0040] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
This study undertook to create small-diameter vascular grafts and assess their structure and mechanical properties to withstand arterial implantation. Twenty samples of intact human internal mammary arteries (IMAs) were collected and decellularized using detergent-based methods. To evaluate residual cellular and extracellular matrix (ECM) components, histological analysis was performed. Moreover, collagen typing and ECM structure were analyzed by Picrosirius red and Movat's pentachrome staining. Scanning electron microscopy was also applied to assess microarchitecture of both endothelial and adventitial surfaces of native and decellularized arterial samples. Furthermore, mechanical tests were performed to evaluate the rigidity and suture strength of the arteries. Human IMAs were completely decellularized in all three segments (proximal, middle, and distal). ECM proteins such as collagen and elastic fibers were efficiently preserved and no structural distortion in intima, media, and adventitial surfaces was observed. The parameters of the mechanical tests revealed no significant differences in the mechanical properties of decellularized arteries in comparison to native arteries with considerable strength, suture retention, and stress relaxation (Young's modulus [MPa] = 0.22 ± 0.023 [native] and 0.22 ± 0.015 [acellular]; and suture strength 0.56 ± 0.19 [native] vs. 0.56 ± 0.12 [acellular], respectively). Decellularized IMA represents a potential arterial scaffold as an alternative to autologous grafts for future arterial bypass surgeries. By this technique, microarchitecture and mechanical integrity of decellularized arteries were considerably similar to native arteries. The goal of this study was to introduce an efficient method for complete decellularization of human IMA and evaluate the ECM and biomechanical properties.
Collapse
Affiliation(s)
- Abdol-Mohammad Kajbafzadeh
- Section of Tissue Engineering and Stem Cell Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Khorramirouz
- Section of Tissue Engineering and Stem Cell Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyede Maryam Kameli
- Section of Tissue Engineering and Stem Cell Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Javad Hashemi
- Section of Tissue Engineering and Stem Cell Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Amin Bagheri
- Section of Tissue Engineering and Stem Cell Therapy, Pediatric Urology and Regenerative Medicine Research Center, Children's Hospital Medical Center, Pediatric Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
45
|
Onwuka E, Best C, Sawyer A, Yi T, Heuer E, Sams M, Wiet M, Zheng H, Kyriakides T, Breuer C. The role of myeloid cell-derived PDGF-B in neotissue formation in a tissue-engineered vascular graft. Regen Med 2017; 12:249-261. [PMID: 28524773 DOI: 10.2217/rme-2016-0141] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
AIM Inflammatory myeloid lineage cells mediate neotissue formation in tissue-engineered vascular grafts, but the molecular mechanism is not completely understood. We examined the role of vasculogenic PDGF-B in tissue-engineered vascular graft neotissue development. MATERIALS & METHODS Myeloid cell-specific PDGF-B knockout mice (PDGF-KO) were generated using bone marrow transplantation, and scaffolds were implanted as inferior vena cava interposition grafts in either PDGF-KO or wild-type mice. RESULTS After 2 weeks, grafts from PDGF-KO mice had more remaining scaffold polymer and less intimal neotissue development. Increased macrophage apoptosis, decreased smooth muscle cell proliferation and decreased collagen content was also observed. CONCLUSION Myeloid cell-derived PDGF contributes to vascular neotissue formation by regulating macrophage apoptosis, smooth muscle cell proliferation and extracellular matrix deposition.
Collapse
Affiliation(s)
- Ekene Onwuka
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Cameron Best
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Andrew Sawyer
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Tai Yi
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Eric Heuer
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Malik Sams
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Matthew Wiet
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Hong Zheng
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Themis Kyriakides
- Vascular Biology & Therapeutics, Yale School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale School of Medicine, New Haven, CT, USA
| | - Christopher Breuer
- Tissue Engineering & Surgical Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, OH, USA.,Department of Pediatric Surgery, Nationwide Children's Hospital, Columbus, OH, USA
| |
Collapse
|
46
|
Manufacturing and Characterization of Novel Electrospun Composite Comprising Polyurethane and Mustard Oil Scaffold with Enhanced Blood Compatibility. Polymers (Basel) 2017; 9:polym9050163. [PMID: 30970842 PMCID: PMC6432176 DOI: 10.3390/polym9050163] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 04/25/2017] [Accepted: 04/28/2017] [Indexed: 11/17/2022] Open
Abstract
The objective of this work is to characterize and investigate the blood compatibility of polyurethane (PU)/mustard oil composites fabricated using electrospinning technique. The fabricated scaffold was characterized using scanning electron microscopy (SEM), Fourier transform infrared spectroscopy (FTIR), atomic force microscopy (AFM), thermogravimetric analysis (TGA) and contact angle measurements. The activated partial thromboplastin time (APPT), prothrombin time (PT) and the hemolytic assay were done to investigate the blood compatibility of the developed composites. The SEM results revealed that the fiber diameter of the composites (761 ± 123 nm) was reduced compared to pristine PU control. The interaction between PU and mustard oil was confirmed by FTIR as evident through the shifting of peaks. The fabricated composites depicted hydrophobic behavior as insinuated by the increase in contact angle measurements. PU/mustard composites displayed improved crystallinity as confirmed by TGA. Atomic force micrographs suggested that developed PU/mustard oil composites showed an increase in the surface roughness (Ra) compared to pure PU. The Ra of pure PU was observed to be 723 nm but for the fabricated PU/mustard oil composite the Ra was found to be 1298 nm (Ra). The hemolytic index value for pure PU and fabricated composites was observed to be 2.73% and 1.15% indicating that developed composites showed a non-hemolytic behavior signifying the safety of the composites with red blood cells. Hence the newly developed composites with improved physicochemical and blood compatibility properties may be considered as a potential candidate for fabricating cardiac patches and grafts.
Collapse
|
47
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide a broad overview of current trends in stem cell research and its applications in cardiovascular medicine. Researches on different stem cell sources, their inherent characteristics, and the limitations they have in medical applications are discussed. Additionally, uses of stem cells for both modeling and treating cardiovascular disease are discussed, taking note of the obstacles these engineered interventions must overcome to be clinically viable. RECENT FINDINGS Tissue engineering aims to replace dysfunctional tissues with engineered constructs. Stem cell technologies have been a great enabling factor in working toward this goal. Many tissue-engineered products are in development that utilize stem cell technology. Although promising, some refinement must be made to these constructs with respect to safety and functionality. A deeper understanding of basic differentiation and tissue developmental mechanisms is required to allow these engineered tissues to be translated into the clinic.
Collapse
Affiliation(s)
- Christopher W Anderson
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Molecular Cell Genetics and Developmental Biology Program, Yale University, New Haven, CT, 06510, USA
| | - Nicole Boardman
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Jiesi Luo
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Jinkyu Park
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA
| | - Yibing Qyang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT, 06510, USA.
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale Cardiovascular Research Center, Yale School of Medicine, 300 George Street, Ste 773A, New Haven, CT, 06511, USA.
- Yale Stem Cell Center, Yale University, New Haven, CT, 06510, USA.
- Department of Pathology, Yale University, New Haven, CT, 06510, USA.
| |
Collapse
|
48
|
Emmert MY, Fioretta ES, Hoerstrup SP. Translational Challenges in Cardiovascular Tissue Engineering. J Cardiovasc Transl Res 2017; 10:139-149. [PMID: 28281240 DOI: 10.1007/s12265-017-9728-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/03/2017] [Indexed: 01/23/2023]
Abstract
Valvular heart disease and congenital heart defects represent a major cause of death around the globe. Although current therapy strategies have rapidly evolved over the decades and are nowadays safe, effective, and applicable to many affected patients, the currently used artificial prostheses are still suboptimal. They do not promote regeneration, physiological remodeling, or growth (particularly important aspects for children) as their native counterparts. This results in the continuous degeneration and subsequent failure of these prostheses which is often associated with an increased morbidity and mortality as well as the need for multiple re-interventions. To overcome this problem, the concept of tissue engineering (TE) has been repeatedly suggested as a potential technology to enable native-like cardiovascular replacements with regenerative and growth capacities, suitable for young adults and children. However, despite promising data from pre-clinical and first clinical pilot trials, the translation and clinical relevance of such TE technologies is still very limited. The reasons that currently limit broad clinical adoption are multifaceted and comprise of scientific, clinical, logistical, technical, and regulatory challenges which need to be overcome. The aim of this review is to provide an overview about the translational problems and challenges in current TE approaches. It further suggests directions and potential solutions on how these issues may be efficiently addressed in the future to accelerate clinical translation. In addition, a particular focus is put on the current regulatory guidelines and the associated challenges for these promising TE technologies.
Collapse
Affiliation(s)
- Maximilian Y Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, Moussonstrasse 13, 8091, Zurich, Switzerland.,Heart Center Zurich, University Hospital Zurich, Zurich, Switzerland.,Wyss Translational Center Zurich, Zurich, Switzerland
| | - Emanuela S Fioretta
- Institute for Regenerative Medicine (IREM), University of Zurich, Moussonstrasse 13, 8091, Zurich, Switzerland
| | - Simon P Hoerstrup
- Institute for Regenerative Medicine (IREM), University of Zurich, Moussonstrasse 13, 8091, Zurich, Switzerland. .,Wyss Translational Center Zurich, Zurich, Switzerland.
| |
Collapse
|
49
|
Shen C, Li Y, Wang H, Meng Q. Mechanically strong interpenetrating network hydrogels for differential cellular adhesion. RSC Adv 2017. [DOI: 10.1039/c7ra01271c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Hydrogels as “soft-and-wet” materials have been widely used as tissue engineering scaffolds due to their similarity to natural extracellular matrix.
Collapse
Affiliation(s)
- Chong Shen
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- PR China
| | - Yuyan Li
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- PR China
| | - Huadi Wang
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- PR China
| | - Qin Meng
- College of Chemical and Biological Engineering
- Zhejiang University
- Hangzhou
- PR China
| |
Collapse
|
50
|
Li C, Wang F, Douglas G, Zhang Z, Guidoin R, Wang L. Comprehensive mechanical characterization of PLA fabric combined with PCL to form a composite structure vascular graft. J Mech Behav Biomed Mater 2016; 69:39-49. [PMID: 28038405 DOI: 10.1016/j.jmbbm.2016.11.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Revised: 11/01/2016] [Accepted: 11/03/2016] [Indexed: 10/20/2022]
Abstract
Vascular grafts made by tissue engineering processes are prone to buckling and twisting, which can impede blood flow and lead to collapse of the vessel. These vascular conduits may suffer not only from insufficient tensile strength, but also from vulnerabilities related to compression, torsion, and pulsatile pressurization. Aiming to develop a tissue engineering-inspired blood conduit, composite vascular graft (cVG) prototypes were created by combining a flexible polylactic acid (PLA) knitted fabric with a soft polycaprolactone (PCL) matrix. The graft is to be populated in-situ with cellular migration and proliferation into the device. Comprehensive characterizations probed the relationship between structure and mechanical properties of the different cVG prototypes. The composite grafts exhibited major improvements in mechanical characteristics compared to single-material devices, with particular improvement in compression and torsional resistance. A commercial expanded polytetrafluoroethylene (ePTFE) vascular graft was used as a control against the proposed composite vascular grafts. CVG devices showed high tensile strength, high bursting strength, and improved suture retention. Compression, elastic recovery, and compliance were similar to those for the ePTFE graft.
Collapse
Affiliation(s)
- Chaojing Li
- Key Laboratory of Textile Science and Technology of Ministry of Education and College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai 201620, China; Department of Surgery, Laval University and Axe of Regenerative Medicine, Research Center CHU, Quebec, Canada
| | - Fujun Wang
- Key Laboratory of Textile Science and Technology of Ministry of Education and College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| | - Graeham Douglas
- Department of Engineering, University of Cambridge, Cambridge CB2 1PZ, UK
| | - Ze Zhang
- Department of Surgery, Laval University and Axe of Regenerative Medicine, Research Center CHU, Quebec, Canada
| | - Robert Guidoin
- Department of Surgery, Laval University and Axe of Regenerative Medicine, Research Center CHU, Quebec, Canada
| | - Lu Wang
- Key Laboratory of Textile Science and Technology of Ministry of Education and College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai 201620, China
| |
Collapse
|